Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 41(12): 111853, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36543145

RESUMO

Here, we ask why the nail base is essential for mammalian digit tip regeneration, focusing on the inductive nail mesenchyme. We identify a transcriptional signature for these cells that includes Lmx1b and show that the Lmx1b-expressing nail mesenchyme is essential for blastema formation. We use a combination of Lmx1bCreERT2-based lineage-tracing and single-cell transcriptional analyses to show that the nail mesenchyme contributes cells for two pro-regenerative mechanisms. One group of cells maintains their identity and regenerates the new nail mesenchyme. A second group contributes specifically to the dorsal blastema, loses their nail mesenchyme phenotype, acquires a blastema transcriptional state that is highly similar to blastema cells of other origins, and ultimately contributes to regeneration of the dorsal but not ventral dermis and bone. Thus, the regenerative necessity for an intact nail base is explained, at least in part, by a requirement for the inductive nail mesenchyme.


Assuntos
Células-Tronco Mesenquimais , Animais , Osso e Ossos , Células Cultivadas , Extremidades , Mamíferos
2.
Cell Rep ; 33(2): 108257, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33053360

RESUMO

Here, we ask how neural stem cells (NSCs) transition in the developing neocortex from a rapidly to a slowly proliferating state, a process required to maintain lifelong stem cell pools. We identify LRIG1, known to regulate receptor tyrosine kinase signaling in other cell types, as a negative regulator of cortical NSC proliferation. LRIG1 is expressed in murine cortical NSCs as they start to proliferate more slowly during embryogenesis and then peaks postnatally when they transition to give rise to a portion of adult NSCs. Constitutive or acute loss of Lrig1 in NSCs over this developmental time frame causes stem cell expansion due to increased proliferation. LRIG1 controls NSC proliferation by associating with and negatively regulating the epidermal growth factor receptor (EGFR). These data support a model in which LRIG1 dampens the stem cell response to EGFR ligands within the cortical environment to slow their proliferation as they transition to postnatal adult NSCs.


Assuntos
Receptores ErbB/metabolismo , Glicoproteínas de Membrana/metabolismo , Neocórtex/citologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Proliferação de Células , Autorrenovação Celular , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Camundongos , Camundongos Knockout , Neurogênese
3.
Nat Med ; 26(8): 1285-1294, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32719487

RESUMO

We asked whether pharmacological stimulation of endogenous neural precursor cells (NPCs) may promote cognitive recovery and brain repair, focusing on the drug metformin, in parallel rodent and human studies of radiation injury. In the rodent cranial radiation model, we found that metformin enhanced the recovery of NPCs in the dentate gyrus, with sex-dependent effects on neurogenesis and cognition. A pilot double-blind, placebo-controlled crossover trial was conducted (ClinicalTrials.gov, NCT02040376) in survivors of pediatric brain tumors who had been treated with cranial radiation. Safety, feasibility, cognitive tests and MRI measures of white matter and the hippocampus were evaluated as endpoints. Twenty-four participants consented and were randomly assigned to complete 12-week cycles of metformin (A) and placebo (B) in either an AB or BA sequence with a 10-week washout period at crossover. Blood draws were conducted to monitor safety. Feasibility was assessed as recruitment rate, medication adherence and procedural adherence. Linear mixed modeling was used to examine cognitive and MRI outcomes as a function of cycle, sequence and treatment. We found no clinically relevant safety concerns and no serious adverse events associated with metformin. Sequence effects were observed for all cognitive outcomes in our linear mixed models. For the subset of participants with complete data in cycle 1, metformin was associated with better performance than placebo on tests of declarative and working memory. We present evidence that a clinical trial examining the effects of metformin on cognition and brain structure is feasible in long-term survivors of pediatric brain tumors and that metformin is safe to use and tolerable in this population. This pilot trial was not intended to test the efficacy of metformin for cognitive recovery and brain growth, but the preliminary results are encouraging and warrant further investigation in a large multicenter phase 3 trial.


Assuntos
Neoplasias Encefálicas/complicações , Disfunção Cognitiva/tratamento farmacológico , Metformina/administração & dosagem , Pediatria/tendências , Adolescente , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Sobreviventes de Câncer , Criança , Pré-Escolar , Cognição/efeitos dos fármacos , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Método Duplo-Cego , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Metformina/efeitos adversos , Neurogênese/efeitos dos fármacos , Projetos Piloto , Resultado do Tratamento , Adulto Jovem
4.
Stem Cell Reports ; 15(1): 140-155, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32559459

RESUMO

Cell transplantation for spinal cord injury (SCI) has largely been studied in sub-acute settings within 1-2 weeks of injury. In contrast, here we transplanted skin-derived precursors differentiated into Schwann cells (SKP-SCs) into the contused rat spinal cord 8 weeks post-injury (wpi). Twenty-one weeks later (29 wpi), SKP-SCs were found to have survived transplantation, integrated with host tissue, and mitigated the formation of a dense glial scar. Furthermore, transplanted SKP-SCs filled much of the lesion sites and greatly enhanced the presence of endogenous SCs, which myelinated thousands of sprouting/spared host axons in and around the injury site. In addition, SKP-SC transplantation improved locomotor outcomes and decreased pathological thickening of bladder wall. To date, functional improvements have very rarely been observed with cell transplantation beyond the sub-acute stage of injury. Hence, these findings indicate that skin-derived SCs are a promising candidate cell type for the treatment of chronic SCI.


Assuntos
Locomoção , Células de Schwann/transplante , Pele/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Bexiga Urinária/patologia , Animais , Axônios/patologia , Doença Crônica , Feminino , Bainha de Mielina/metabolismo , Regeneração Nervosa , Neuroglia/patologia , Ratos Sprague-Dawley , Medula Espinal/patologia , Medula Espinal/fisiopatologia
5.
Dev Cell ; 52(4): 509-524.e9, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-31902657

RESUMO

Here, we investigate the origin and nature of blastema cells that regenerate the adult murine digit tip. We show that Pdgfra-expressing mesenchymal cells in uninjured digits establish the regenerative blastema and are essential for regeneration. Single-cell profiling shows that the mesenchymal blastema cells are distinct from both uninjured digit and embryonic limb or digit Pdgfra-positive cells. This unique blastema state is environmentally determined; dermal fibroblasts transplanted into the regenerative, but not non-regenerative, digit express blastema-state genes and contribute to bone regeneration. Moreover, lineage tracing with single-cell profiling indicates that endogenous osteoblasts or osteocytes acquire a blastema mesenchymal transcriptional state and contribute to both dermis and bone regeneration. Thus, mammalian digit tip regeneration occurs via a distinct adult mechanism where the regenerative environment promotes acquisition of a blastema state that enables cells from tissues such as bone to contribute to the regeneration of other mesenchymal tissues such as the dermis.


Assuntos
Diferenciação Celular , Extremidades/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Mesenquimais/citologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Regeneração , Animais , Linhagem da Célula , Células Cultivadas , Extremidades/embriologia , Extremidades/lesões , Feminino , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Análise de Célula Única , Transcriptoma
6.
Cell Stem Cell ; 24(2): 240-256.e9, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30503141

RESUMO

Peripheral innervation plays an important role in regulating tissue repair and regeneration. Here we provide evidence that injured peripheral nerves provide a reservoir of mesenchymal precursor cells that can directly contribute to murine digit tip regeneration and skin repair. In particular, using single-cell RNA sequencing and lineage tracing, we identify transcriptionally distinct mesenchymal cell populations within the control and injured adult nerve, including neural crest-derived cells in the endoneurium with characteristics of mesenchymal precursor cells. Culture and transplantation studies show that these nerve-derived mesenchymal cells have the potential to differentiate into non-nerve lineages. Moreover, following digit tip amputation, neural crest-derived nerve mesenchymal cells contribute to the regenerative blastema and, ultimately, to the regenerated bone. Similarly, neural crest-derived nerve mesenchymal cells contribute to the dermis during skin wound healing. These findings support a model where peripheral nerves directly contribute mesenchymal precursor cells to promote repair and regeneration of injured mammalian tissues.


Assuntos
Células-Tronco Mesenquimais/citologia , Regeneração Nervosa/fisiologia , Tecido Nervoso/patologia , Cicatrização , Animais , Regeneração Óssea , Diferenciação Celular , Linhagem da Célula , Camundongos , Crista Neural/citologia , Osteogênese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Células de Schwann/patologia , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Transcrição Gênica , Transcriptoma/genética
7.
Stem Cell Reports ; 10(5): 1464-1480, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29628394

RESUMO

Circulating systemic factors can regulate adult neural stem cell (NSC) biology, but the identity of these circulating cues is still being defined. Here, we have focused on the cytokine interleukin-6 (IL-6), since increased circulating levels of IL-6 are associated with neural pathologies such as autism and bipolar disorder. We show that IL-6 promotes proliferation of post-natal murine forebrain NSCs and that, when the IL-6 receptor is inducibly knocked out in post-natal or adult neural precursors, this causes a long-term decrease in forebrain NSCs. Moreover, a transient circulating surge of IL-6 in perinatal or adult mice causes an acute increase in neural precursor proliferation followed by long-term depletion of adult NSC pools. Thus, IL-6 signaling is both necessary and sufficient for adult NSC self-renewal, and acute perturbations in circulating IL-6, as observed in many pathological situations, have long-lasting effects on the size of adult NSC pools.


Assuntos
Células-Tronco Adultas/citologia , Crescimento e Desenvolvimento , Interleucina-6/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/metabolismo , Animais , Animais Recém-Nascidos , Contagem de Células , Proliferação de Células , Interleucina-6/sangue , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Fatores de Tempo
8.
J Cell Biol ; 216(11): 3655-3675, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28877995

RESUMO

Axon degeneration is an early event and pathological in neurodegenerative conditions and nerve injuries. To discover agents that suppress neuronal death and axonal degeneration, we performed drug screens on primary rodent neurons and identified the pan-kinase inhibitor foretinib, which potently rescued sympathetic, sensory, and motor wt and SOD1 mutant neurons from trophic factor withdrawal-induced degeneration. By using primary sympathetic neurons grown in mass cultures and Campenot chambers, we show that foretinib protected neurons by suppressing both known degenerative pathways and a new pathway involving unliganded TrkA and transcriptional regulation of the proapoptotic BH3 family members BimEL, Harakiri,and Puma, culminating in preservation of mitochondria in the degenerative setting. Foretinib delayed chemotherapy-induced and Wallerian axonal degeneration in culture by preventing axotomy-induced local energy deficit and preserving mitochondria, and peripheral Wallerian degeneration in vivo. These findings identify a new axon degeneration pathway and a potentially clinically useful therapeutic drug.


Assuntos
Anilidas/farmacologia , Lesões por Esmagamento/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinolinas/farmacologia , Receptor trkA/antagonistas & inibidores , Nervo Isquiático/efeitos dos fármacos , Neuropatia Ciática/tratamento farmacológico , Degeneração Walleriana , Fibras Adrenérgicas/efeitos dos fármacos , Fibras Adrenérgicas/enzimologia , Fibras Adrenérgicas/patologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Axônios/efeitos dos fármacos , Axônios/enzimologia , Axônios/patologia , Células Cultivadas , Lesões por Esmagamento/enzimologia , Lesões por Esmagamento/genética , Lesões por Esmagamento/patologia , Citoproteção , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Genótipo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/enzimologia , Neurônios Motores/patologia , Mutação , Neurônios/enzimologia , Neurônios/patologia , Fenótipo , Fosforilação , Ratos Sprague-Dawley , Receptor trkA/genética , Receptor trkA/metabolismo , Nervo Isquiático/enzimologia , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Neuropatia Ciática/enzimologia , Neuropatia Ciática/genética , Neuropatia Ciática/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/enzimologia , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Fatores de Tempo , Transcrição Gênica
9.
Cell Stem Cell ; 19(4): 433-448, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27376984

RESUMO

Adult mammals have lost multi-tissue regenerative capacity, except for the distal digit, which is able to regenerate via mechanisms that remain largely unknown. Here, we show that, after adult mouse distal digit removal, nerve-associated Schwann cell precursors (SCPs) dedifferentiate and secrete growth factors that promote expansion of the blastema and digit regeneration. When SCPs were dysregulated or ablated, mesenchymal precursor proliferation in the blastema was decreased and nail and bone regeneration were impaired. Transplantation of exogenous SCPs rescued these regeneration defects. We found that SCPs secrete factors that promote self-renewal of mesenchymal precursors, and we used transcriptomic and proteomic analysis to define candidate factors. Two of these, oncostatin M (OSM) and platelet-derived growth factor AA (PDGF-AA), are made by SCPs in the regenerating digit and rescued the deficits in regeneration caused by loss of SCPs. As all peripheral tissues contain nerves, these results could have broad implications for mammalian tissue repair and regeneration.


Assuntos
Desdiferenciação Celular , Extremidades/fisiologia , Mamíferos/fisiologia , Células-Tronco Neurais/citologia , Comunicação Parácrina , Regeneração , Células de Schwann/citologia , Envelhecimento/fisiologia , Animais , Desdiferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Denervação , Extremidades/inervação , Deleção de Genes , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mesoderma/citologia , Camundongos , Camundongos Knockout , Células-Tronco Neurais/transplante , Oncostatina M/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Regeneração/efeitos dos fármacos , Fatores de Transcrição SOXB1/metabolismo , Células de Schwann/transplante , Pele/patologia , Cicatrização/efeitos dos fármacos
10.
Stem Cell Reports ; 5(6): 988-995, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26677765

RESUMO

The recruitment of endogenous adult neural stem cells for brain repair is a promising regenerative therapeutic strategy. This strategy involves stimulation of multiple stages of adult neural stem cell development, including proliferation, self-renewal, and differentiation. Currently, there is a lack of a single therapeutic approach that can act on these multiple stages of adult neural stem cell development to enhance neural regeneration. Here we show that metformin, an FDA-approved diabetes drug, promotes proliferation, self-renewal, and differentiation of adult neural precursors (NPCs). Specifically, we show that metformin enhances adult NPC proliferation and self-renewal dependent upon the p53 family member and transcription factor TAp73, while it promotes neuronal differentiation of these cells by activating the AMPK-aPKC-CBP pathway. Thus, metformin represents an optimal candidate neuro-regenerative agent that is capable of not only expanding the adult NPC population but also subsequently driving them toward neuronal differentiation by activating two distinct molecular pathways.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Proteína de Ligação a CREB/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/metabolismo , Proteína Quinase C/metabolismo , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/metabolismo
11.
J Neurosci ; 34(15): 5164-75, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24719096

RESUMO

The Snail transcription factor plays a key role in regulating diverse developmental processes but is not thought to play a role in mammalian neural precursors. Here, we have examined radial glial precursor cells of the embryonic murine cortex and demonstrate that Snail regulates their survival, self-renewal, and differentiation into intermediate progenitors and neurons via two distinct and separable target pathways. First, Snail promotes cell survival by antagonizing a p53-dependent death pathway because coincident p53 knockdown rescues survival deficits caused by Snail knockdown. Second, we show that the cell cycle phosphatase Cdc25b is regulated by Snail in radial precursors and that Cdc25b coexpression is sufficient to rescue the decreased radial precursor proliferation and differentiation observed upon Snail knockdown. Thus, Snail acts via p53 and Cdc25b to coordinately regulate multiple aspects of mammalian embryonic neural precursor biology.


Assuntos
Córtex Cerebral/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Células Ependimogliais/citologia , Células Ependimogliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Células-Tronco Neurais/citologia , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo
12.
Cell Stem Cell ; 13(5): 564-76, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24209760

RESUMO

The mechanisms that regulate the establishment of adult stem cell pools during normal and perturbed mammalian development are still largely unknown. Here, we asked whether a maternal cytokine surge, which occurs during human maternal infections and has been implicated in cognitive disorders, might have long-lasting consequences for neural stem cell pools in adult progeny. We show that transient, maternally administered interleukin-6 (IL-6) resulted in an expanded adult forebrain neural precursor pool and perturbed olfactory neurogenesis in offspring months after fetal exposure. This increase is likely the long-term consequence of acute hyperactivation of an endogenous autocrine/paracrine IL-6-dependent self-renewal pathway that normally regulates the number of forebrain neural precursors. These studies therefore identify an IL-6-dependent neural stem cell self-renewal pathway in vivo, and support a model in which transiently increased maternal cytokines can act through this pathway in offspring to deregulate neural precursor biology from embryogenesis throughout life.


Assuntos
Interleucina-6/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Interleucina-6/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
13.
J Neurosci ; 33(31): 12569-85, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23904595

RESUMO

The molecular mechanisms that regulate adult neural precursor cell (NPC) survival, and thus maintain adult neurogenesis, are not well defined. Here, we investigate the role of p63, a p53 family member, in adult NPC function in mice. Conditional ablation of p63 in adult NPCs or p63 haploinsufficiency led to reduced numbers of NPCs and newborn neurons in the neurogenic zones of the hippocampus and lateral ventricles and in the olfactory bulb. These reductions were attributable to enhanced apoptosis of NPCs and newborn neurons and were rescued by inhibition of caspase activity, p53, or the p53 apoptotic effector PUMA (p53-upregulated modulator of apoptosis). Moreover, these cellular deficits were functionally important because they led to perturbations in hippocampus-dependent memory formation. These results indicate that p63 regulates the numbers of adult NPCs and adult-born neurons as well as neural stem cell-dependent cognitive functions, and that it does so, at least in part, by inhibiting p53-dependent cell death.


Assuntos
Células-Tronco Adultas/fisiologia , Comportamento Exploratório/fisiologia , Hipocampo/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Ventrículos Cerebrais/citologia , Condicionamento Psicológico/fisiologia , Sinais (Psicologia) , Comportamento Exploratório/efeitos dos fármacos , Medo/psicologia , Proteínas de Filamentos Intermediários/genética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Nestina , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Fosfoproteínas/genética , Proteínas/genética , RNA não Traduzido , Tamoxifeno/farmacologia , Transativadores/genética , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
14.
Neurobiol Aging ; 34(2): 387-99, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22592019

RESUMO

Haploinsufficiency for the p53 family member p73 causes behavioral and neuroanatomical correlates of neurodegeneration in aging mice, including the appearance of aberrant phospho-tau-positive aggregates. Here, we show that these aggregates and tau hyperphosphorylation, as well as a generalized dysregulation of the tau kinases GSK3ß, c-Abl, and Cdk5, occur in the brains of aged p73+/- mice. To investigate whether p73 haploinsufficiency therefore represents a general risk factor for tau hyperphosphorylation during neurodegeneration, we crossed the p73+/- mice with 2 mouse models of neurodegeneration, TgCRND8+/Ø mice that express human mutant amyloid precursor protein, and Pin1-/- mice. We show that haploinsufficiency for p73 leads to the early appearance of phospho-tau-positive aggregates, tau hyperphosphorylation, and activation of GSK3ß, c-Abl, and Cdk5 in the brains of both of these mouse models. Moreover, p73+/-;TgCRND8+/Ø mice display a shortened lifespan relative to TgCRND8+/Ø mice that are wild type for p73. Thus, p73 is required to protect the murine brain from tau hyperphosphorylation during aging and degeneration.


Assuntos
Envelhecimento/genética , Doença de Alzheimer/genética , Encéfalo/metabolismo , Proteínas de Ligação a DNA/genética , Haploinsuficiência , Proteínas Nucleares/genética , Proteínas Supressoras de Tumor/genética , Proteínas tau/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Animais , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Genes abl/genética , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Longevidade/genética , Camundongos , Proteínas Nucleares/metabolismo , Fosforilação , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 31(12): 2938-48, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21852558

RESUMO

OBJECTIVE: The goal of this study was to characterize the factors and conditions required for smooth muscle cell (SMC)-directed differentiation of Sox2(+) multipotent rat and human skin-derived precursors (SKPs) and to define whether they represent a source of fully functional vascular SMCs for applications in vivo. METHODS AND RESULTS: We found that rat SKPs can differentiate almost exclusively into SMCs by reducing serum concentrations to 0.5% to 2% and plating them at low density. Human SKPs derived from foreskin required the addition of transforming growth factor-ß1 or -ß3 to differentiate into SMCs, but they did so even in the absence of serum. SMC formation was confirmed by quantitative reverse transcription-polymerase chain reaction, immunocytochemistry, and fluorescence-activated cell sorting, with increased expression of smoothelin-B and little to no expression of telokin or smooth muscle γ-actin, together indicating that SKPs differentiated into vascular rather than visceral SMCs. Rat and human SKP-derived SMCs were able to contract in vitro and also wrap around and support new capillary and larger blood vessel formation in angiogenesis assays in vivo. CONCLUSIONS: SKPs are Sox2(+) progenitors that represent an attainable autologous source of stem cells that can be easily differentiated into functional vascular SMCs in defined serum-free conditions without reprogramming. SKPs represent a clinically viable cell source for potential therapeutic applications in neovascularization.


Assuntos
Diferenciação Celular , Células-Tronco Multipotentes/citologia , Músculo Liso Vascular/citologia , Pele/citologia , Actinas/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Humanos , Masculino , Modelos Animais , Células-Tronco Multipotentes/efeitos dos fármacos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Neovascularização Fisiológica/fisiologia , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta3/farmacologia
16.
J Neurosci Res ; 89(3): 286-98, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21259316

RESUMO

Microglia play important roles in the damaged or degenerating adult nervous system. However, the role of microglia in embryonic brain development is still largely uncharacterized. Here we show that microglia are present in regions of the developing brain that contain neural precursors from E11 onward. To determine whether these microglia are important for neural precursor maintenance or self-renewal, we cultured embryonic neural precursors from the cortex of PU.1(-/-) mice, which we show lack resident microglia during embryogenesis. Cell survival and neurogenesis were similar in cultures from PU.1(-/-) vs. PU.1(+/+) mice, but precursor proliferation and astrogenesis were both reduced. Cortical precursors depleted of microglia also displayed decreased precursor proliferation and astrogenesis, and these deficits could be rescued when microglia were added back to the cultures. Moreover, when the number of microglia present in cortical precursor cultures was increased above normal levels, astrogenesis but not neurogenesis was increased. Together these results demonstrate that microglia present within the embryonic neural precursor niche can regulate neural precursor development and suggest that alterations in microglial number as a consequence of genetic or pathological events could perturb neural development by directly affecting embryonic neural precursors.


Assuntos
Diferenciação Celular/genética , Córtex Cerebral/anatomia & histologia , Ventrículos Cerebrais/citologia , Microglia/fisiologia , Células-Tronco/fisiologia , Fatores Etários , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Células Cultivadas , Córtex Cerebral/embriologia , Embrião de Mamíferos , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Filamentos Intermediários/metabolismo , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese/genética , Neurônios/fisiologia , Antígenos O/genética , Proteínas Proto-Oncogênicas/deficiência , Transativadores/deficiência , Tubulina (Proteína)/metabolismo
17.
Dev Biol ; 347(2): 348-59, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20832397

RESUMO

Coffin-Lowry Syndrome (CLS) is an X-linked genetic disorder associated with cognitive and behavioural impairments. CLS patients present with loss-of-function mutations in the RPS6KA3 gene encoding the mitogen-activated protein kinase (MAPK)-activated kinase p90 ribosomal S6 kinase 2 (Rsk2). Although Rsk2 is expressed in the embryonic brain, its function remains largely uncharacterized. To this end, we isolated murine cortical precursors at embryonic day 12 (E12), a timepoint when neuronal differentiation is initiated, and knocked-down Rsk2 expression levels using shRNA. We performed similar experiments in vivo using in utero electroporations to express shRNA against Rsk2. Rsk2 knockdown resulted in a significant decrease in neurogenesis and an increase in the proportion of proliferating Pax6-positive radial precursor cells, indicating that Rsk2 is essential for cortical radial precursors to differentiate into neurons. In contrast, reducing Rsk2 levels in vitro or in vivo had no effect on the generation of astrocytes. Thus, Rsk2 loss-of-function, as seen in CLS, perturbs the differentiation of neural precursors into neurons, and maintains them instead as proliferating radial precursor cells, a defect that may underlie the cognitive dysfunction seen in CLS.


Assuntos
Síndrome de Coffin-Lowry/etiologia , Neurogênese/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Animais , Sequência de Bases , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/enzimologia , Síndrome de Coffin-Lowry/embriologia , Síndrome de Coffin-Lowry/enzimologia , Síndrome de Coffin-Lowry/genética , Modelos Animais de Doenças , Células-Tronco Embrionárias , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Neurogênese/genética , Gravidez , RNA Interferente Pequeno/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/genética
18.
EMBO Mol Med ; 2(9): 371-84, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20721990

RESUMO

Neuroblastoma (NB) is the most deadly extra-cranial solid tumour in children necessitating an urgent need for effective and less toxic treatments. One reason for the lack of efficacious treatments may be the inability of existing drugs to target the tumour-initiating or cancer stem cell population responsible for sustaining tumour growth, metastases and relapse. Here, we describe a strategy to identify compounds that selectively target patient-derived cancer stem cell-like tumour-initiating cells (TICs) while sparing normal paediatric stem cells (skin-derived precursors, SKPs) and characterize two therapeutic candidates. DECA-14 and rapamycin were identified as NB TIC-selective agents. Both compounds induced TIC death at nanomolar concentrations in vitro, significantly reduced NB xenograft tumour weight in vivo, and dramatically decreased self-renewal or tumour-initiation capacity in treated tumours. These results demonstrate that differential drug sensitivities between TICs and normal paediatric stem cells can be exploited to identify novel, patient-specific and potentially less toxic therapies.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Dequalínio/análogos & derivados , Células-Tronco Neoplásicas/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Sirolimo/uso terapêutico , Bibliotecas de Moléculas Pequenas/química , Animais , Apoptose , Dequalínio/química , Dequalínio/uso terapêutico , Transporte de Elétrons , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Mitocôndrias/genética , Mitocôndrias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neuroblastoma/genética , Bibliotecas de Moléculas Pequenas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Genes Dev ; 24(6): 549-60, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20194434

RESUMO

Mice with a complete deficiency of p73 have severe neurological and immunological defects due to the absence of all TAp73 and DeltaNp73 isoforms. As part of our ongoing program to distinguish the biological functions of these isoforms, we generated mice that are selectively deficient for the DeltaNp73 isoform. Mice lacking DeltaNp73 (DeltaNp73(-/-) mice) are viable and fertile but display signs of neurodegeneration. Cells from DeltaNp73(-/-) mice are sensitized to DNA-damaging agents and show an increase in p53-dependent apoptosis. When analyzing the DNA damage response (DDR) in DeltaNp73(-/-) cells, we discovered a completely new role for DeltaNp73 in inhibiting the molecular signal emanating from a DNA break to the DDR pathway. We found that DeltaNp73 localizes directly to the site of DNA damage, can interact with the DNA damage sensor protein 53BP1, and inhibits ATM activation and subsequent p53 phosphorylation. This novel finding may explain why human tumors with high levels of DeltaNp73 expression show enhanced resistance to chemotherapy.


Assuntos
Dano ao DNA , Reparo do DNA/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transdução de Sinais , Animais , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia , Encéfalo/patologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Feminino , Fertilidade/genética , Regulação da Expressão Gênica , Células HCT116 , Humanos , Longevidade/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Neurodegenerativas/genética , Fosforilação , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
20.
Nat Neurosci ; 13(5): 559-66, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20348920

RESUMO

Axonal degeneration is important during development but has not been thought to function in the intact mature nervous system. Here, we provide evidence that degeneration of adult axons occurs in the intact rodent brain through a p75 neurotrophin receptor (p75NTR)- and myelin-dependent mechanism. Specifically, we show that p75NTR-mediated axonal degeneration prevents septal cholinergic axons from aberrantly growing onto myelinated tracts in vivo or on a myelin substrate in culture. Myelin also triggers local degeneration of p75NTR-expressing sympathetic axons that is rescued by increasing TrkA signaling or elevating intracellular cyclic AMP. Myelin-mediated degeneration occurs when neurotrophins bind to p75NTR, and involves p75NTR-dependent sequestration of Rho guanine nucleotide dissociation inhibitor (Rho-GDI). Moreover, degeneration, but not growth inhibition, requires downstream activation of Rho and caspase-6. These data indicate that p75NTR maintains the specificity of neural connectivity by preventing inappropriate sprouting onto myelinated tracts and provide a physiological explanation for myelin inhibition after neural injury.


Assuntos
Axônios/fisiologia , Bainha de Mielina/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Caspase 6/metabolismo , Células Cultivadas , Colina O-Acetiltransferase/metabolismo , Cofilina 1/metabolismo , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Fator de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/metabolismo , Neurônios/fisiologia , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor de Fator de Crescimento Neural/genética , Receptor trkA/metabolismo , Septo do Cérebro/citologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Medula Espinal/metabolismo , Fatores de Tempo , Tubulina (Proteína)/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA